251
|
Tunici P, Irvin D, Liu G, Yuan X, Zhaohui Z, Ng H, Yu JS. Brain tumor stem cells: new targets for clinical treatments? Neurosurg Focus 2006; 20:E27. [PMID: 16709033 DOI: 10.3171/foc.2006.20.4.17] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
✓ The observation of similarities between the self-renewal mechanisms of stem cells and cancer cells has led to the new concept of the cancer stem cell. In cases of leukemia, multiple myeloma, and breast cancer, cells with a high self-renewal potential have been identified. Furthermore, investigators have shown these cells' ability to drive the formation and growth of the tumor. Brain tumors have also been reported to possess a subpopulation of cancer stemlike cells that have the ability to proliferate, self-renew, and be multipotent. When grafted into mice, these cells are also able to generate a tumor that recapitulates that of the patient from whom the cells were derived. The identification and characterization of this new category of cells call for new therapies capable of selectively targeting and killing these multifaceted cells.
Collapse
Affiliation(s)
- Patrizia Tunici
- Maxine Dunitz Neurosurgical Institute, Los Angeles, California 90048, USA
| | | | | | | | | | | | | |
Collapse
|
252
|
Zhou JX, Jia LW, Liu WM, Miao CL, Liu S, Cao YJ, Duan EK. Role of sonic hedgehog in maintaining a pool of proliferating stem cells in the human fetal epidermis. Hum Reprod 2006; 21:1698-704. [PMID: 16571640 DOI: 10.1093/humrep/del086] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The mammalian epidermis is maintained by the ongoing proliferation of a subpopulation of keratinocytes known as epidermal stem cells. Sonic hedgehog (Shh) can regulate morphogenesis of hair follicles and several types of skin cancer, but the effect of Shh on proliferation of human putative epidermal stem cells (HPESCs) is poorly understood. METHODS AND RESULTS We first found that Shh, its receptors Patched1 (Ptc1) as well as Smoothened (Smo) and its downstream transcription factor Gli-1 were expressed in the basal layer of human fetal epidermis and freshly sorted HPESCs. Next, treatment of HPESCs with media conditioned by Shh-N-expressing cells promoted cell proliferation, whereas inhibition of Shh by cyclopamine, a specific inhibitor of Shh signalling, had an opposite effect. Interestingly, the mitogenic effect of epidermal growth factor (EGF) on HPESCs was efficiently abolished by cyclopamine. Finally, bone morphogenetic protein 4 (BMP-4), a potential downstream effector of Shh signalling, increased HPESC proliferation in a concentration-dependent manner. CONCLUSIONS Shh is an important regulator of HPESC proliferation in the basal layer of human fetal epidermis and modulates the cell responsiveness to EGF, which will assist to unravel the mechanisms that regulate stem cell proliferation and neoplasia in the human epidermis.
Collapse
Affiliation(s)
- Jia-xi Zhou
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
253
|
Bénazéraf B, Chen Q, Peco E, Lobjois V, Médevielle F, Ducommun B, Pituello F. Identification of an unexpected link between the Shh pathway and a G2/M regulator, the phosphatase CDC25B. Dev Biol 2006; 294:133-47. [PMID: 16564519 DOI: 10.1016/j.ydbio.2006.02.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2005] [Revised: 01/12/2006] [Accepted: 02/22/2006] [Indexed: 12/23/2022]
Abstract
Sonic hedgehog (Shh) signaling controls numerous aspects of vertebrate development, including proliferation of precursors in different organs. Identification of molecules that link the Shh pathway to cell cycle machinery is therefore of major importance for an understanding of the mechanisms underlying Shh-dependent proliferation. Here, we show that an actor in the control of entry into mitosis, the phosphatase CDC25B, is transcriptionally upregulated by the Shh/Gli pathway. Unlike other G2/M regulators, CDC25B is highly expressed in domains of Shh activity, including the ventral neural tube and the posterior limb bud. Loss- and gain-of-function experiments reveal that Shh contributes to CDC25B transcriptional activation in the neural tube both of chick and mouse embryos. Moreover, CDC25B transcripts are absent from the posterior limb bud of Shh-/- mice, while anterior grafts of Shh-expressing cells in the chicken limb bud induce ectopic CDC25B expression. Arresting the cell cycle does not reduce the level of CDC25B expression in the neural tube strongly suggesting that the upregulation of CDC25B is not an indirect consequence of the Shh-dependent proliferation. These data reveal an unexpected developmental link between the Shh pathway and a participant in G2/M control.
Collapse
Affiliation(s)
- Bertrand Bénazéraf
- Centre de Biologie du Développement, UMR5547 CNRS-Université P. Sabatier, IFR109--Institut d'Exploration Fonctionnelle des Génomes, 118 route de Narbonne, 31062 Toulouse Cedex, France
| | | | | | | | | | | | | |
Collapse
|
254
|
Lien WH, Klezovitch O, Fernandez TE, Delrow J, Vasioukhin V. alphaE-catenin controls cerebral cortical size by regulating the hedgehog signaling pathway. Science 2006; 311:1609-12. [PMID: 16543460 PMCID: PMC2556178 DOI: 10.1126/science.1121449] [Citation(s) in RCA: 212] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
During development, cells monitor and adjust their rates of accumulation to produce organs of predetermined size. We show here that central nervous system-specific deletion of the essential adherens junction gene, alphaE-catenin, causes abnormal activation of the hedgehog pathway, resulting in shortening of the cell cycle, decreased apoptosis, and cortical hyperplasia. We propose that alphaE-catenin connects cell-density-dependent adherens junctions with the developmental hedgehog pathway and that this connection may provide a negative feedback loop controlling the size of developing cerebral cortex.
Collapse
Affiliation(s)
- Wen-Hui Lien
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, 98195
| | - Olga Klezovitch
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
| | - Tania E. Fernandez
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
| | - Jeff Delrow
- Genomics Resource, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
| | - Valeri Vasioukhin
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109
| |
Collapse
|
255
|
Datta MW, Hernandez AM, Schlicht MJ, Kahler AJ, DeGueme AM, Dhir R, Shah RB, Farach-Carson C, Barrett A, Datta S. Perlecan, a candidate gene for the CAPB locus, regulates prostate cancer cell growth via the Sonic Hedgehog pathway. Mol Cancer 2006; 5:9. [PMID: 16507112 PMCID: PMC1421430 DOI: 10.1186/1476-4598-5-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2005] [Accepted: 03/01/2006] [Indexed: 12/24/2022] Open
Abstract
Background Genetic studies associated the CAPB locus with familial risk of brain and prostate cancers. We have identified HSPG2 (Perlecan) as a candidate gene for CAPB. Previously we have linked Perlecan to Hedgehog signaling in Drosophila. More recently, we have demonstrated the importance of Hedgehog signaling in humans for advanced prostate cancer. Results Here we demonstrate Perlecan expression in prostate cancer, and its function in prostate cancer cell growth through interaction and modulation of Sonic Hedgehog (SHH) signaling. Perlecan expression in prostate cancer tissues correlates with a high Gleason score and rapid cell proliferation. Perlecan is highly expressed in prostate cancer cell lines, including androgen insensitive cell lines and cell lines selected for metastatic properties. Inhibition of Perlecan expression in these cell lines decreases cell growth. Simultaneous blockade of Perlecan expression and androgen signaling in the androgen-sensitive cell line LNCaP was additive, indicating the independence of these two pathways. Perlecan expression correlates with SHH in tumor tissue microarrays and increased tumor cell proliferation based on Ki-67 immunohistochemistry. Inhibition of Perlecan expression by siRNA in prostate cancer cell lines decreases SHH signaling while expression of the downstream SHH effector GLI1 rescues the proliferation defect. Perlecan forms complexes with increasing amounts of SHH that correlate with increasing metastatic potential of the prostate cancer cell line. SHH signaling also increases in the more metastatic cell lines. Metastatic prostate cancer cell lines grown under serum-starved conditions (low androgen and growth factors) resulted in maintenance of Perlecan expression. Under low androgen, low growth factor conditions, Perlecan expression level correlates with the ability of the cells to maintain SHH signaling. Conclusion We have demonstrated that Perlecan, a candidate gene for the CAPB locus, is a new component of the SHH pathway in prostate tumors and works independently of androgen signaling. In metastatic tumor cells increased SHH signaling correlates with the maintenance of Perlecan expression and more Perlecan-SHH complexes. Perlecan is a proteoglycan that regulates extracellular and stromal accessibility to growth factors such as SHH, thus allowing for the maintenance of SHH signaling under growth factor limiting conditions. This proteoglycan represents an important central regulator of SHH activity and presents an ideal drug target for blocking SHH effects.
Collapse
Affiliation(s)
- Milton W Datta
- Departments of Pathology and Urology, Emory University, Atlanta, GA, 30322, USA
| | - Ana Maria Hernandez
- Department of Biochemistry and Biophysics, Texas A&M University, 2128 TAMU, College Station, Texas 77843-2128, USA
| | - Michael J Schlicht
- Departments of Pathology and Urology, Emory University, Atlanta, GA, 30322, USA
| | - Andrea J Kahler
- Departments of Pathology and Urology, Emory University, Atlanta, GA, 30322, USA
| | - Amy M DeGueme
- Departments of Pathology and Urology, Emory University, Atlanta, GA, 30322, USA
| | - Rajiv Dhir
- Department of Pathology, University of Pittsburgh Medical Center, 200 Lothrop Street, Pittsburgh PA, 15232, USA
| | - Rajal B Shah
- Department of Pathology, University of Michigan, 1500 Catherine Ave, Ann Arbor, MI, 48109, USA
| | - Cindy Farach-Carson
- Department of Biological Sciences, University of Delaware, 304 Wolfe Hall, Newark, DE, 19716, USA
| | - Andrea Barrett
- Department of Biochemistry and Biophysics, Texas A&M University, 2128 TAMU, College Station, Texas 77843-2128, USA
| | - Sumana Datta
- Department of Biochemistry and Biophysics, Texas A&M University, 2128 TAMU, College Station, Texas 77843-2128, USA
| |
Collapse
|
256
|
Mimeault M, Moore E, Moniaux N, Hénichart JP, Depreux P, Lin MF, Batra SK. Cytotoxic effects induced by a combination of cyclopamine and gefitinib, the selective hedgehog and epidermal growth factor receptor signaling inhibitors, in prostate cancer cells. Int J Cancer 2006; 118:1022-31. [PMID: 16108016 DOI: 10.1002/ijc.21440] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Although the blockade of the hedgehog cascade by using cyclopamine has been reported to inhibit the growth of some cancer cell types, few studies on the mechanism by which this drug alone or in combination with other cytotoxic agents induces its cytotoxic effect have been reported. In our study, we evaluate, for the first time, the antiproliferative and cytotoxic effects induced by a combination of selective SMO inhibitor, cyclopamine and epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, gefitinib on metastatic prostate cancer (PC) cells. The results revealed that cyclopamine, alone or at a lower concentration in combination with gefitinib, inhibited the growth of sonic hedgehog- (SHH), epidermal growth factor- (EGF) and serum-stimulated androgen-sensitive LNCaP-C33 and LNCaP-LN3 and androgen-independent LNCaP-C81, DU145 and PC3 cells. The antiproliferative effect of cyclopamine and gefitinib, alone or in combination, was mediated via a blockade of the PC3 cells in the G1 phase of the cell cycle. Importantly, the combined cyclopamine and gefitinib also caused a higher rate of apoptotic death of PC cells compared to single agents. The cytotoxic effect induced by these drugs in PC3 cells appears to be mediated at least, in part, via the mitochondrial pathway through the depolarization of the mitochondrial membrane and the release of cytochrome c and reactive oxygen species into the cytosol. This was also accompanied by the activation of caspase cascades, PARP cleavage and DNA fragmentation. Additionally, the combined cyclopamine and gefitinib were more effective at suppressing the invasiveness of PC3 cells through matrigel in vitro as the drugs alone. These findings indicate that the simultaneous blockade of SHH-GLI-1 and EGF-EGFR signaling, which results in the growth arrest and massive rate of apoptotic cell death, represents a promising strategy for a more effective treatment of metastatic PC forms.
Collapse
Affiliation(s)
- Murielle Mimeault
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, College of Medicine, Omaha, NE 68198-5870, USA
| | | | | | | | | | | | | |
Collapse
|
257
|
Cutcliffe C, Kersey D, Huang CC, Zeng Y, Walterhouse D, Perlman EJ. Clear cell sarcoma of the kidney: up-regulation of neural markers with activation of the sonic hedgehog and Akt pathways. Clin Cancer Res 2006; 11:7986-94. [PMID: 16299227 DOI: 10.1158/1078-0432.ccr-05-1354] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE AND EXPERIMENTAL DESIGN Clear cell sarcoma of the kidney (CCSK), the second most common renal tumor in children, poses significant diagnostic challenges. No positive diagnostic markers are available, and the pathogenesis of CCSK remains an enigma. To address these challenges, the gene expression patterns of 14 CCSKs were compared with 15 Wilms tumors and 3 fetal kidney samples using oligonucleotide arrays. RESULTS Using unsupervised methods, the gene expression profile of CCSK was distinctive: differentially expressed genes could largely be grouped into four categories: (a) a wide variety of neural markers, (b) members of the Sonic hedgehog pathway, (c) members of the phosphoinositide-3-kinase/Akt cell proliferation pathway, and (d) known therapeutic targets. Corresponding changes in critical proteins using Western blot and/or immunohistochemistry confirmed the up-regulation of these pathways and proteins. In particular, CD117 and epidermal growth factor receptor are up-regulated at the protein level in many CCSKs, providing potential therapeutic targets. One of the neural markers, nerve growth factor receptor, represents a promising diagnostic tool for CCSK. CONCLUSIONS This study suggests that CCSKs arise within a renal mesenchymal cell that shows a wide variety of neural markers. As such, it seems to be susceptible to genetic changes also seen in a variety of other neuroectodermal and neuronal tumors, including activation of Sonic hedgehog and phosphoinositide-3-kinase/Akt pathways. Involvement of these pathways in CCSKs implicates their widening role in tumorigenesis.
Collapse
Affiliation(s)
- Colleen Cutcliffe
- Department of Pathology and Laboratory Medicine, Division of Hematology/Oncology, Children's Memorial Hospital, Chicago, Illinois 60614, USA
| | | | | | | | | | | |
Collapse
|
258
|
Chen BY, Lin DPC, Liu JY, Chang H, Huang PH, Chen YL, Chang HH. A mouse prostate cancer model induced by Hedgehog overexpression. J Biomed Sci 2005; 13:373-84. [PMID: 16374524 DOI: 10.1007/s11373-005-9050-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2005] [Accepted: 11/28/2005] [Indexed: 10/25/2022] Open
Abstract
Hedgehog is a regulatory protein during embryonic development and its abnormal activation in adult tissues has been implicated in tumorigenesis within sites where epithelial-mesenchymal interactions take place. In the prostate, Hedgehog signaling activation was observed during advanced cancer progression and metastasis, but whether Hedgehog overexpression can initiate prostate tumorigenesis remains unknown. We introduced a Hedgehog-expressing vector by intra-prostate injection and electroporation to address the effects of Hedgehog overexpression. The manipulation caused lesions with characteristic prostatic intraepithelial neoplasia or even prostatic cancer (CaP) phenotypes within 30 days, with Hedgehog overexpression demonstrated by immunohistochemistry and Western blot detections. The tumorigenic phenotypes were confirmed by discontinuity of basal cell marker p63, mix-up of CK-8/CK-18 positive epithelial cells in the stoma as well as absence of alpha-SMA positive fibro-muscular sheath. Comparable Hedgehog overexpression was found in human CaP specimen. Thus, Hedgehog overexpression induced prostate tumorigenesis starting from the normal status. Furthermore, a mouse prostate cancer model induced by Hedgehog overexpression was established and may be used for testing novel therapeutical approaches targeting at Hedgehog signaling pathway.
Collapse
Affiliation(s)
- Bo-Yie Chen
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
259
|
Ohta M, Tateishi K, Kanai F, Watabe H, Kondo S, Guleng B, Tanaka Y, Asaoka Y, Jazag A, Imamura J, Ijichi H, Ikenoue T, Sata M, Miyagishi M, Taira K, Tada M, Kawabe T, Omata M. p53-Independent negative regulation of p21/cyclin-dependent kinase-interacting protein 1 by the sonic hedgehog-glioma-associated oncogene 1 pathway in gastric carcinoma cells. Cancer Res 2005; 65:10822-10829. [PMID: 16322228 DOI: 10.1158/0008-5472.can-05-0777] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The activation of Hedgehog (Hh) signaling has been implicated in the growth of various tumor types, including gastric carcinoma. However, the precise mechanisms of Hh activation and suppression of tumor growth by the blockade of Hh signaling in gastric carcinoma cells remain unknown. The aim of this study was to elucidate the mechanism of abnormal Hh signaling and the key molecules contributing to dysregulated growth of gastric carcinoma. The Sonic hedgehog (Shh) ligand and its receptor Patched were expressed in all five gastric carcinoma cell lines examined (MKN1, MKN7, MKN45, MKN74, and AGS cells). The blockade of Hh signaling with anti-Shh antibody inhibited the growth of all five gastric carcinoma cell lines. Shh was overexpressed (mean, 12.8-fold) in 8 of 14 (57.0%) cancerous tissue samples from patients with gastric carcinoma as compared with expression in the surrounding noncancerous tissues. The disruption of glioma-associated oncogene 1 (Gli1) by small interfering RNA induced an increase in p21/cyclin-dependent kinase-interacting protein 1 (CIP1), interfered with the G1-S transition, and suppressed cell proliferation. The stimulation or inhibition of Hh signaling did not affect p53 activity and the induction of p21/CIP1 expression and the G1 arrest by inhibition of Hh signaling were not affected by the p53 status. These findings suggest that the overexpression of Shh contributes to constitutive Hh activation and that this signaling pathway negatively regulates p21/CIP1 through a Gli1-dependent and p53-independent mechanism in gastric carcinoma cells.
Collapse
Affiliation(s)
- Miki Ohta
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Hongo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
260
|
Das AV, Edakkot S, Thoreson WB, James J, Bhattacharya S, Ahmad I. Membrane properties of retinal stem cells/progenitors. Prog Retin Eye Res 2005; 24:663-81. [PMID: 15939659 DOI: 10.1016/j.preteyeres.2005.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The membrane properties of cells help integrate extrinsic information relayed through growth factors, chemokines, extracellular matrix, gap junctions and neurotransmitters towards modulating cell-intrinsic properties, which in turn determine whether cells remain quiescent, proliferate, differentiate, establish contact with other cells or remove themselves by activating programmed cell death. This review highlights some of the membrane properties of early and late retinal stem cells/progenitors, which are likely to be helpful in the identification and enrichment of these cells and in understanding mechanisms underlying their maintenance and differentiation. Understanding of membrane properties of retinal stem cells/progenitors is essential for the successful formulation of approaches to treat retinal degeneration and diseases by cell therapy.
Collapse
Affiliation(s)
- Ani V Das
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198-7691, USA
| | | | | | | | | | | |
Collapse
|
261
|
Hara Y, Katow H. Exclusive expression of hedgehog in small micromere descendants during early embryogenesis in the sea urchin, Hemicentrotus pulcherrimus. Gene Expr Patterns 2005; 5:503-10. [PMID: 15749078 DOI: 10.1016/j.modgep.2004.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2004] [Revised: 11/26/2004] [Accepted: 12/13/2004] [Indexed: 11/17/2022]
Abstract
Hedgehog (hh) is a multifunctional extracellular protein, and known as an essential signal molecule in morphogenetic movement in animal embryos. We have cloned, sequenced, and studied dynamic localization of Hphh, a hedgehog homologue of the sea urchin, Hemicentrotus pulcherrimus. The origin of Hphh transcribing cells was also verified during early embryogenesis. The amino acid sequence of Hphh shows high homology to Lvhh, an hh homologue cloned in the sea urchin, Lytechinus variegatus. Reverse transcriptase polymerase chain reaction showed that the transcription of Hphh occurred at and after 19 h post-fertilization (19 hpf) mesenchyme blastula stage until, at least, 69 hpf 4-arm pluteus stage. Whole mount in situ hybridization showed Hphh transcription sites in a few cells at the tip of archenteron in 30 hpf gastrulae. At around 45 hpf 2-arm pluteus stage, the number of Hphh transcribed cells was 8, and unequally split to two groups, 5 cells in left coelomic sac and 3 cells in right coelomic sac. A cell lineage tracing by staining the small micromeres with 5-Bromo-2-deoxyuridine showed that Hphh was transcribed exclusively in all the small micromere descendants and comprised the coelomic sacs in 69 hpf plutei.
Collapse
Affiliation(s)
- Yoshinobu Hara
- Laboratory of Developmental and Cell Biology, Graduate School of Life Sciences, Tohoku University, Asamushi, Aomori 039-3501, Japan
| | | |
Collapse
|
262
|
Piedimonte LR, Wailes IK, Weiner HL. Medulloblastoma: mouse models and novel targeted therapies based on the Sonic hedgehog pathway. Neurosurg Focus 2005; 19:E8. [PMID: 16398472 DOI: 10.3171/foc.2005.19.5.9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Understanding molecular pathways, signaling cascades, and genetic alterations activated during tumorigenesis is essential for the development of targeted cancer treatments. In children, tumors of the central nervous system are thought to arise from progenitor cells that show considerable temporal and spatial heterogeneity in a developmental environment that is different from that of the adult. Investigating the molecular basis of pediatric tumors is critical because it is likely to generate novel treatments. Animal models have brought many important advances in this field. In this review the authors discuss the mouse models based on the Sonic hedgehog pathway, which have provided a better knowledge of the genetic and molecular alterations of medulloblastoma.
Collapse
Affiliation(s)
- Leandro R Piedimonte
- Division of Pediatric Neurosurgery, Department of Neurosurgery, New York University School of Medicine, New York, New York 10016, USA
| | | | | |
Collapse
|
263
|
Li MH, Bouffet E, Hawkins CE, Squire JA, Huang A. Molecular genetics of supratentorial primitive neuroectodermal tumors and pineoblastoma. Neurosurg Focus 2005; 19:E3. [PMID: 16398467 DOI: 10.3171/foc.2005.19.5.4] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The supratentorial primitive neuroectodermal tumors (PNETs) are a group of highly malignant lesions primarily affecting young children. Although these tumors are histologically indistinguishable from infratentorial medulloblastoma, they often respond poorly to medulloblastoma-specific therapy. Indeed, existing molecular genetic studies indicate that supratentorial PNETs have transcriptional and cytogenetic profiles that are different from those of medullo-blastomas, thus pointing to unique biological derivation for the supratentorial PNET. Due to the rarity of these tumors and disagreement about their histopathological diagnoses, very little is known about the molecular characteristics of the supratentorial PNET. Clearly, future concerted efforts to characterize the molecular features of these rare tumors will be necessary for development of more effective supratentorial PNET treatment protocols and appropriate disease models. In this article the authors review existing molecular genetic data derived from human and mouse studies, with the aim of providing some insight into the putative histogenesis of these rare tumors and the underlying transforming pathways that drive their development. Studies of the related but distinct pineoblastoma PNET are also reviewed.
Collapse
Affiliation(s)
- Mei Hua Li
- Arthur and Sonia Labatt Brain Tumor Research Centre, Cancer Research Program, Division of Hematology and Oncology, Department of Pediatric Laboratory Medicine, Hospital for Sick Children, Toronto, Canada
| | | | | | | | | |
Collapse
|
264
|
Abstract
The entire vertebrate nervous system develops from a simple epithelial sheet, the neural plate which, along development, acquires the large number and wide variety of neuronal cell types required for the construction of a functional mature nervous system. These include processes of growth and pattern formation of the neural tube that are achieved through complicated and tightly regulated genetic interactions. Pattern formation, particularly in the vertebrate central nervous system, is one of the best examples of a morphogen-type of function. Cell cycle progression, however, is generally accepted to be dependent on cell-intrinsic factors. Recent studies have demonstrated that proliferation of neural precursors is also somehow controlled by secreted signaling molecules, well-known by their role as morphogens, such as fibroblast growth factor (FGF), vertebrate orthologs of the Drosophila wingless (Wnt), hedgehog (Hh), and transforming growth factor beta (TGF-beta) families, that in turn regulate the activity of factors controlling cell cycle progression. In this review we will summarize the experimental data that support the idea that classical morphogens can be reused to regulate proliferation of neural precursors.
Collapse
Affiliation(s)
- Jordi Cayuso
- Instituto de Biología Molecular de Barcelona (CSIC), Parc Cientific de Barcelona, C/ Josep Samitier 1-5, Barcelona 08028, Spain
| | | |
Collapse
|
265
|
Stecca B, Ruiz i Altaba A. Brain as a paradigm of organ growth: Hedgehog-Gli signaling in neural stem cells and brain tumors. ACTA ACUST UNITED AC 2005; 64:476-90. [PMID: 16041742 DOI: 10.1002/neu.20160] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The Hedgehog-Gli (Hh-Gli) signaling pathway is essential for numerous events during the development of many animal cell types and organs. In particular, it controls neural cell precursor proliferation in dorsal brain structures and regulates the number of neural stem cells in distinct embryonic, perinatal, and adult niches, such as the developing neocortex, the subventricular zone of the lateral ventricle of the forebrain, and the hippocampus. We have proposed that Hh-Gli signaling regulates dorsal brain growth during ontogeny and that its differential regulation underlays evolutionary change in the morphology (size and shape) of dorsal brain structures. It is also critically involved in sporadic brain tumorigenesis--as well as several other human cancer--suggesting that tumors derive from stem cells or progenitors maintaining an inappropriate active Hh-Gli pathway. Importantly, we and others have demonstrated that human sporadic tumors from the brain and other organs require sustained HH-GLI signaling for sustained growth and survival. Modulating HH-GLI signaling thus represents a novel rational avenue to treat, on one hand, brain degeneration and injury by inducing controlled HH-GLI-mediated regeneration and growth, and on the other hand, to combat cancer by blocking its abnormal activity in tumor cells.
Collapse
Affiliation(s)
- Barbara Stecca
- Department of Genetic Medicine and Development, University of Geneva Medical School, 8242 CMU, 1 rue Michel Servet, CH-1211 Geneva 4, Switzerland
| | | |
Collapse
|
266
|
Fogarty MP, Kessler JD, Wechsler-Reya RJ. Morphing into cancer: the role of developmental signaling pathways in brain tumor formation. ACTA ACUST UNITED AC 2005; 64:458-75. [PMID: 16041741 DOI: 10.1002/neu.20166] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Morphogens play a critical role in most aspects of development, including expansion and patterning of the central nervous system. Activating germline mutations in components of the Hedgehog and Wnt pathways have provided evidence for the important roles morphogens play in the genesis of brain tumors such as cerebellar medulloblastoma. In addition, aberrant expression of transforming growth factor-beta (TGF-beta) superfamily members has been demonstrated to contribute to progression of malignant gliomas. This review summarizes our current knowledge about the roles of morphogens in central nervous system tumorigenesis.
Collapse
Affiliation(s)
- Marie P Fogarty
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | |
Collapse
|
267
|
Vieira C, Garda AL, Shimamura K, Martinez S. Thalamic development induced by Shh in the chick embryo. Dev Biol 2005; 284:351-63. [PMID: 16026780 DOI: 10.1016/j.ydbio.2005.05.031] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2005] [Revised: 05/23/2005] [Accepted: 05/24/2005] [Indexed: 12/19/2022]
Abstract
Patterning of the early neural tube is achieved in part by the inductive signals, which arise from neuroepithelial signaling centers. The zona limitans intrathalamica (ZLI) is a neuroepithelial domain in the alar plate of the diencephalon which separates the prethalamus from the thalamus. The ZLI has recently been considered to be a possible secondary organizer, effecting its inductions via sonic hedgehog (Shh), a signaling molecule which drives morphogenetic information for the thalamus. Using experimental embryological techniques involving the generation of chimeric embryos, we show that the formation of the ZLI in the diencephalic alar plate is due to an interaction between the prechordal and epichordal plate neuroepithelia. We also provide evidence that Shh expression in the ZLI underlies the morphogenetic activity of this putative diencephalic organizer. Ectopic Shh led to the auto-induction of its own gene expression in host cells, as well as to the expression of other genes involved in diencephalic regionalization and histogenesis. Analysis of long-term surviving embryos after Shh ectopic expression demonstrated that Shh was able to induce thalamic structures and local overgrowth. Overall, these results indicate that Shh expressed in the ZLI plays an important role in diencephalic growth and in the development of the thalamus.
Collapse
Affiliation(s)
- Claudia Vieira
- Neuroscience Institute, Miguel Hernandez University N-332, Km 87, E-03550 Alicante, Spain
| | | | | | | |
Collapse
|
268
|
Merchant M, Evangelista M, Luoh SM, Frantz GD, Chalasani S, Carano RAD, van Hoy M, Ramirez J, Ogasawara AK, McFarland LM, Filvaroff EH, French DM, de Sauvage FJ. Loss of the serine/threonine kinase fused results in postnatal growth defects and lethality due to progressive hydrocephalus. Mol Cell Biol 2005; 25:7054-68. [PMID: 16055717 PMCID: PMC1190232 DOI: 10.1128/mcb.25.16.7054-7068.2005] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The Drosophila Fused (Fu) kinase is an integral component of the Hedgehog (Hh) pathway that helps promote Hh-dependent gene transcription. Vertebrate homologues of Fu function in the Hh pathway in vitro, suggesting that Fu is evolutionarily conserved. We have generated fused (stk36) knockout mice to address the in vivo function of the mouse Fu (mFu) homologue. fused knockouts develop normally, being born in Mendelian ratios, but fail to thrive within 2 weeks, displaying profound growth retardation with communicating hydrocephalus and early mortality. The fused gene is expressed highly in ependymal cells and the choroid plexus, tissues involved in the production and circulation of cerebral spinal fluid (CSF), suggesting that loss of mFu disrupts CSF homeostasis. Similarly, fused is highly expressed in the nasal epithelium, where fused knockouts display bilateral suppurative rhinitis. No obvious defects were observed in the development of organs where Hh signaling is required (limbs, face, bones, etc.). Specification of neuronal cell fates by Hh in the neural tube was normal in fused knockouts, and induction of Hh target genes in numerous tissues is not affected by the loss of mFu. Furthermore, stimulation of fused knockout cerebellar granule cells to proliferate with Sonic Hh revealed no defect in Hh signal transmission. These results show that the mFu homologue is not required for Hh signaling during embryonic development but is required for proper postnatal development, possibly by regulating the CSF homeostasis or ciliary function.
Collapse
Affiliation(s)
- Mark Merchant
- Department of Molecular Biology, South San Francisco, CA 94080, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
269
|
Abstract
Transforming growth factor beta (TGF-beta) signaling leads to a number of biological end points involving cell growth, differentiation, and morphogenesis. Typically, the cellular effect accompanies an induction of mesodermal cell fate and inhibition of neural cell differentiation. However, during pathological conditions, these defined effects of TGF-beta can be reversed; for example, the growth-inhibitory effect is replaced with its tumor promoting ability. A multitude of factors and cross-signaling pathways have been reported to be involved in modulating the dual effects of TGF-beta. In this review, we focus on the potential role of TGF-beta signal transduction during development of neural progenitor cells and its relation to glioblastoma development from neural stem cells.
Collapse
Affiliation(s)
- Nady Golestaneh
- School of Medicine, Georgetown University, 3900 Reservoir Road NW, Washington, DC 20007, USA
| | | |
Collapse
|
270
|
Affiliation(s)
- Nader Sanai
- Department of Neurological Surgery, Brain Tumor Research Center, and the Developmental Stem Cell Biology Program, University of California, San Francisco, CA 94143, USA.
| | | | | |
Collapse
|
271
|
Koudijs MJ, den Broeder MJ, Keijser A, Wienholds E, Houwing S, van Rooijen EMHC, Geisler R, van Eeden FJM. The zebrafish mutants dre, uki, and lep encode negative regulators of the hedgehog signaling pathway. PLoS Genet 2005; 1:e19. [PMID: 16121254 PMCID: PMC1189072 DOI: 10.1371/journal.pgen.0010019] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2005] [Accepted: 06/23/2005] [Indexed: 11/19/2022] Open
Abstract
Proliferation is one of the basic processes that control embryogenesis. To identify factors involved in the regulation of proliferation, we performed a zebrafish genetic screen in which we used proliferating cell nuclear antigen (PCNA) expression as a readout. Two mutants, hu418B and hu540A, show increased PCNA expression. Morphologically both mutants resembled the dre (dreumes), uki (ukkie), and lep (leprechaun) mutant class and both are shown to be additional uki alleles. Surprisingly, although an increased size is detected of multiple structures in these mutant embryos, adults become dwarfs. We show that these mutations disrupt repressors of the Hedgehog (Hh) signaling pathway. The dre, uki, and lep loci encode Su(fu) (suppressor of fused), Hip (Hedgehog interacting protein), and Ptc2 (Patched2) proteins, respectively. This class of mutants is therefore unique compared to previously described Hh mutants from zebrafish genetic screens, which mainly show loss of Hh signaling. Furthermore, su(fu) and ptc2 mutants have not been described in vertebrate model systems before. Inhibiting Hh activity by cyclopamine rescues uki and lep mutants and confirms the overactivation of the Hh signaling pathway in these mutants. Triple uki/dre/lep mutants show neither an additive increase in PCNA expression nor enhanced embryonic phenotypes, suggesting that other negative regulators, possibly Ptc1, prevent further activation of the Hh signaling pathway. The effects of increased Hh signaling resulting from the genetic alterations in the uki, dre, and lep mutants differ from phenotypes described as a result of Hh overexpression and therefore provide additional insight into the role of Hh signaling during vertebrate development. In a screen aimed at finding genes that control proliferation in the zebrafish embryo, three mutants were identified. Mutants showed an increase in size of several structures including the brain, the retina, and the fins. Surprisingly, although size was increased in the embryos, adults remained small. Cloning of these genes revealed that increased Hedgehog signaling was at the basis of the phenotype, because all three genes encoded known repressors of the Hedgehog signaling pathway: Ptc2, Su(Fu), and Hip. Hedgehog is known to play a role in proliferation. For instance, ectopic Hedgehog signaling can lead to several tumors including basal cell carcinoma and medulloblastoma. However, the phenotypes were still a surprise, because earlier experiments in zebrafish embryos suggested that activation should lead to patterning rather than proliferation defects. Current models of the pathway predict that these genes act independently to inhibit the signal but curiously, redundancy amongst these genes was not found, because triple mutants looked like the single mutants. The conclusion is that weak activation of Hedgehog signaling can already lead to stimulation of growth in the absence of patterning defects, and that the Hedgehog signal is probably kept in check by the last inhibitor: Ptc1. A mutant for the ptc1 gene has recently been created and will put the model to the test.
Collapse
Affiliation(s)
- Marco J Koudijs
- Hubrecht Laboratory, The Netherlands Institute for Developmental Biology, Utrecht, The Netherlands
| | - Marjo J. den Broeder
- Hubrecht Laboratory, The Netherlands Institute for Developmental Biology, Utrecht, The Netherlands
| | - Astrid Keijser
- Hubrecht Laboratory, The Netherlands Institute for Developmental Biology, Utrecht, The Netherlands
| | - Erno Wienholds
- Hubrecht Laboratory, The Netherlands Institute for Developmental Biology, Utrecht, The Netherlands
| | - Saskia Houwing
- Hubrecht Laboratory, The Netherlands Institute for Developmental Biology, Utrecht, The Netherlands
| | | | - Robert Geisler
- Max-Planck-Institut für Entwicklungsbiologie, Tübingen, Germany
| | - Fredericus J. M. van Eeden
- Hubrecht Laboratory, The Netherlands Institute for Developmental Biology, Utrecht, The Netherlands
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
272
|
Stecca B, Mas C, Ruiz i Altaba A. Interference with HH-GLI signaling inhibits prostate cancer. Trends Mol Med 2005; 11:199-203. [PMID: 15882606 DOI: 10.1016/j.molmed.2005.03.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The Hedgehog-Gli (Hh-Gli) signaling pathway controls many aspects of tissue patterning, cell proliferation, differentiation and regeneration and regulates cell number in various organs. In adults, the Hh-Gli pathway remains active in a number of stem cells and regenerating tissues. Inappropriate and uncontrolled HH-GLI pathway activation has been demonstrated in a variety of human cancers. Three recent papers show that components of the pathway are expressed in human prostate tumors and, more importantly, that prostate cancers depend on sustained HH-GLI signaling. These data raise the possibility of a new therapeutic approach to treat this often lethal disease.
Collapse
Affiliation(s)
- Barbara Stecca
- Department of Genetic Medicine and Development, University of Geneva Medical School, 8242 CMU, 1 rue Michel Servet, CH-1211 Geneva 4, Switzerland
| | | | | |
Collapse
|
273
|
Sanchez P, Ruiz i Altaba A. In vivo inhibition of endogenous brain tumors through systemic interference of Hedgehog signaling in mice. Mech Dev 2005; 122:223-30. [PMID: 15652709 DOI: 10.1016/j.mod.2004.10.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2004] [Revised: 10/05/2004] [Accepted: 10/05/2004] [Indexed: 10/26/2022]
Abstract
The full spectrum of developmental potential includes normal as well as abnormal and disease states. We therefore subscribe to the idea that tumors derive from the operation of paradevelopmental programs that yield consistent and recognizable morphologies. Work in frogs and mice shows that Hedgehog (Hh)-Gli signaling controls stem cell lineages and that its deregulation leads to tumor formation. Moreover, human tumor cells require sustained Hh-Gli signaling for proliferation as cyclopamine, an alkaloid of the lily Veratrum californicum that blocks the Hh pathway, inhibits the growth of different tumor cells in vitro as well as in subcutaneous xenografts. However, the evidence that systemic treatment is an effective anti-cancer therapy is missing. Here we have used Ptc1(+/-); p53(-/-) mice which develop medulloblastoma to test the ability of cyclopamine to inhibit endogenous tumor growth in vivo after tumor initiation through intraperitoneal delivery, which avoids the brain damage associated with direct injection. We find that systemic cyclopamine administration improves the health of Ptc1(+/-);p53(-/-) animals. Analyses of the cerebella of cyclopamine-treated animals show a severe reduction in tumor size and a large decrease in the number of Ptc1-expressing cells, as a readout of cells with an active Hu-Gli pathway, as well as an impairment of their proliferative capacity, always in comparison with vehicle treated mice. Our data demonstrate that systemic treatment with cyclopamine inhibits tumor growth in the brain supporting its therapeutical value for human HH-dependent tumors. They also demonstrate that even the complete loss of the well-known tumor suppressor p53 does not render the tumor independent of Hh pathway function.
Collapse
Affiliation(s)
- Pilar Sanchez
- The Skirball Institute, NYU School of Medicine, 540 First Avenue, New York, NY 10012, USA
| | | |
Collapse
|
274
|
Sanchez P, Clement V, Ruiz i Altaba A. Therapeutic targeting of the Hedgehog-GLI pathway in prostate cancer. Cancer Res 2005; 65:2990-2. [PMID: 15833820 DOI: 10.1158/0008-5472.can-05-0439] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The Hedgehog-GLI signaling pathway is important in animal development and tumorigenesis. Recent findings indicate that the growth and survival of human prostate cancer cells rely upon sustained signaling from the Hedgehog-GLI pathway. These findings have prompted a novel rational strategy for therapeutic treatment of prostate tumors, including metastatic tumors.
Collapse
Affiliation(s)
- Pilar Sanchez
- Department of Genetic Medicine and Development, University of Geneva Medical School, CH-1211 Geneva 4, Switzerland
| | | | | |
Collapse
|
275
|
Abstract
Prostate cancer is the most common malignancy and the second leading cancer-related cause of death in men in the USA. Despite enormous efforts in understanding the molecular basis of prostate cancer, very little progress has been made in prevention and treatment of this often lethal cancer. Recent studies have demonstrated that hedgehog signaling is frequently activated in advanced or metastatic prostate cancers. With small molecule inhibitors available to analyze the hedgehog signaling pathway, a novel rationale for prostate cancer therapy can be devised.
Collapse
Affiliation(s)
- Jingwu Xie
- University of Texas Medical Branch at Galveston, Sealy Centers for Cancer Cell Biology and Environmental Health, Department of Pharmacology and Toxicology, 301 University Blvd, Galveston, TX 77555-1048, USA.
| |
Collapse
|
276
|
Rao G, Pedone CA, Del Valle L, Reiss K, Holland EC, Fults DW. Sonic hedgehog and insulin-like growth factor signaling synergize to induce medulloblastoma formation from nestin-expressing neural progenitors in mice. Oncogene 2005; 23:6156-62. [PMID: 15195141 DOI: 10.1038/sj.onc.1207818] [Citation(s) in RCA: 180] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Medulloblastoma (MB) is a malignant brain tumor that arises in the cerebellum of children. Activation of the Sonic hedgehog/Patched (Shh/Ptc) signaling pathway in neural progenitor cells of the cerebellum induces MBs in mice. The incomplete penetrance of tumor formation in mice, coupled with the low frequency of mutations in Shh/Ptc pathway genes in human tumors, suggests that other signaling molecules cooperate with Shh to enhance MB formation. We modeled the ability of insulin-like growth factor (IGF) signaling to induce MB using the RCAS/tv-a system, which allows postnatal gene transfer and expression in a cell-type-specific manner. We used RCAS retroviral vectors to target expression of Shh, IGF2, and activated Akt to nestin-expressing neural progenitors in the cerebella of newborn mice. The incidence of Shh-induced tumor formation (15%) was enhanced by coexpression with IGF2 (39%) and Akt (48%). Neither IGF2 nor Akt caused tumors when expressed independently. The induced tumors showed upregulated expression of insulin receptor substrate 1 and phosphorylated forms of IGF1 receptor and Akt, mimicking activated IGF signaling found in human MBs. These results indicate that combined activation of the Shh/Ptc and IGF signaling pathways is an important mechanism in MB pathogenesis.
Collapse
Affiliation(s)
- Ganesh Rao
- Department of Neurosurgery, University of Utah School of Medicine, 30 North 1900 East, Salt Lake City, UT 84132-2303, USA
| | | | | | | | | | | |
Collapse
|
277
|
Lefort K, Dotto GP. Notch signaling in the integrated control of keratinocyte growth/differentiation and tumor suppression. Semin Cancer Biol 2005; 14:374-86. [PMID: 15288263 DOI: 10.1016/j.semcancer.2004.04.017] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Oncogenesis is closely linked to abnormalities in cell differentiation. Notch signaling provides an important form of intercellular communication involved in cell fate determination, stem cell potential and differentiation. Here we review the role of this pathway in the integrated growth/differentiation control of the keratinocyte cell type, and the maintenance of normal skin homeostasis. In parallel with the pro-differentiation function of Notch1 in keratinocytes, we discuss recent evidence pointing to a tumor suppressor function of this gene in both mouse skin and human cervical carcinogenesis. The possibility that Notch signaling elicits signals with a duality of growth positive and negative function will be discussed.
Collapse
Affiliation(s)
- Karine Lefort
- Department of Biochemistry, Lausanne University, Chemin des Boveresses 155, CH-1066 Epalinges, Switzerland
| | | |
Collapse
|
278
|
Vestergaard J, Bak M, Larsen LA. The hedgehog signaling pathway in cancer. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2005; 40:1-28. [PMID: 17153478 DOI: 10.1007/3-540-27671-8_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Affiliation(s)
- Janni Vestergaard
- Wilhelm Johannsen Centre for Functional Genome Research, Department of Medical Biochemistry and Genetics, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | | | | |
Collapse
|
279
|
Abstract
During the last century, experiments on the chick embryo established that the ballooning expansion of the early forebrain and midbrain vesicles is dependent on the underlying axial (notochordal) mesoderm. Transient separation of the early midbrain primordium from the notochord causes subsequent collapse of both midbrain and forebrain (telencephalic) vesicles, accompanied by pronounced folding of the neural epithelium. More recent experiments have shown that vesicle collapse is caused by defective Sonic Hedgehog (Shh) signaling from the notochord and floor plate. Separation of the notochord from the brain causes loss of ventral Shh expression, resulting in reduced cell proliferation and increased cell death in the expanding neural epithelium, and culminating in vesicle collapse. These experiments are reviewed here, and set in the context of other studies illustrating the wide range of molecular and cellular processes that cause abnormal brain morphogenesis when perturbed. We also speculate that variation in the regulation of signaling pathways such as Hedgehog may have played a significant part in generating rapid morphogenetic changes during the evolution of the vertebrate brain.
Collapse
Affiliation(s)
- David Tannahill
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
| | | | | |
Collapse
|
280
|
Moshiri A, McGuire CR, Reh TA. Sonic hedgehog regulates proliferation of the retinal ciliary marginal zone in posthatch chicks. Dev Dyn 2005; 233:66-75. [PMID: 15759272 DOI: 10.1002/dvdy.20299] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The ciliary marginal zone (CMZ) has long been known to be a source of postembryonic neuronal production in the retinas of fish and amphibians, and more recently, birds. However, there is little known about the factors that are required for the maintenance of this neural stem cell zone. The cells of the CMZ respond to mitogens such as endothelial growth factor, insulin-like growth factor-1, and insulin, factors that are also mitogenic for embryonic retinal progenitors, suggesting that the continued expression of embryonic mitogenic factors might be required to maintain the postembryonic proliferative potential of the CMZ. To test this hypothesis, we examined the expression and functional role of a critical embryonic retinal progenitor mitogen, Sonic hedgehog (Shh) in the regulation of proliferation of the cells of the CMZ. We have found that Shh is concentrated at the retinal margin of postembryonic chicks. Moreover, we report that intraocular injection of Shh stimulates proliferation of the CMZ cells, whereas cyclopamine, an inhibitor of the Shh pathway, inhibits CMZ proliferation. We conclude that Shh signaling is an important factor in the maintenance of postembryonic retinal neurogenesis.
Collapse
Affiliation(s)
- Ala Moshiri
- Neurobiology and Behavior Program, Department of Biological Structure, University of Washington, School of Medicine, Seattle, WA 98195, USA
| | | | | |
Collapse
|
281
|
Abstract
Cancer is increasingly being viewed as a stem cell disease, both in its propagation by a minority of cells with stem-cell-like properties and in its possible derivation from normal tissue stem cells. But stem cell activity is tightly controlled, raising the question of how normal regulation might be subverted in carcinogenesis. The long-known association between cancer and chronic tissue injury, and the more recently appreciated roles of Hedgehog and Wnt signalling pathways in tissue regeneration, stem cell renewal and cancer growth together suggest that carcinogenesis proceeds by misappropriating homeostatic mechanisms that govern tissue repair and stem cell self-renewal.
Collapse
Affiliation(s)
- Philip A Beachy
- Department of Molecular Biology and Genetics, The Howard Hughes Medical Institute, Baltimore, Maryland 21205, USA.
| | | | | |
Collapse
|
282
|
Katano M. Hedgehog signaling pathway as a therapeutic target in breast cancer. Cancer Lett 2004; 227:99-104. [PMID: 16112412 DOI: 10.1016/j.canlet.2004.11.030] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2004] [Accepted: 11/17/2004] [Indexed: 12/19/2022]
Abstract
The Hedgehog (Hh) signaling pathway, which is well conserved even in mammals and other vertebrate species, has long been known to direct growth and patterning during embryonic development. It has been shown that the Hh pathway also plays a critical role in mouse normal mammary gland development. Namely, it has been shown that disruption of the Hh pathway-related genes such as Patched-1 and Gli2 leads to ductal dysplasias that closely resemble some hyperplasia of human breast. In addition, it has been reported that breast carcinoma cells have disruption of these genes. These findings strongly indicate a contribution of the Hh pathway to development of human breast carcinoma. In fact, constitutive activation of the Hh pathway was found in most of 52 surgically resected breast carcinoma specimens. Interestingly, exposure to cyclopamine, a steroidal alkaroid that blocks the Hh pathway, suppressed the growth of the Hh pathway-activated breast carcinoma cells. Thus, the Hh pathway may function in progression of breast carcinoma. In this short review, possibilities of the Hh pathway as a new therapeutic target in breast carcinoma will be mainly discussed.
Collapse
Affiliation(s)
- Mitsuo Katano
- Department of Cancer Therapy and Research, Graduate School of Medical Siences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
283
|
Hallahan AR, Pritchard JI, Hansen S, Benson M, Stoeck J, Hatton BA, Russell TL, Ellenbogen RG, Bernstein ID, Beachy PA, Olson JM. The SmoA1 Mouse Model Reveals That Notch Signaling Is Critical for the Growth and Survival of Sonic Hedgehog-Induced Medulloblastomas. Cancer Res 2004; 64:7794-800. [PMID: 15520185 DOI: 10.1158/0008-5472.can-04-1813] [Citation(s) in RCA: 329] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To develop a genetically faithful model of medulloblastoma with increased tumor incidence compared with the current best model we activated the Sonic Hedgehog (Shh) pathway by transgenically expressing a constitutively active form of Smoothened in mouse cerebellar granule neuron precursors (ND2:SmoA1 mice). This resulted in early cerebellar granule cell hyper-proliferation and a 48% incidence of medulloblastoma formation. Gene expression studies showed an increase in the known Shh targets Gli1 and Nmyc that correlated with increasing hyperplasia and tumor formation. Notch2 and the Notch target gene, HES5, were also significantly elevated in Smoothened-induced tumors showing that Shh pathway activation is sufficient to induce Notch pathway signaling. In human medulloblastomas reverse transcription-PCR for Shh and Notch targets revealed activation of both of these pathways in most tumors when compared with normal cerebellum. Notch pathway inhibition with soluble Delta ligand or gamma secretase inhibitors resulted in a marked reduction of viable cell numbers in medulloblastoma cell lines and primary tumor cultures. Treatment of mice with D283 medulloblastoma xenografts with a gamma secretase inhibitor resulted in decreased proliferation and increased apoptosis, confirming that Notch signaling contributes to human medulloblastoma proliferation and survival. Medulloblastomas in ND2:SmoA1 mice and humans have concomitant increase in Shh and Notch pathway activities, both of which contribute to tumor survival.
Collapse
Affiliation(s)
- Andrew R Hallahan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
284
|
|
285
|
Abstract
Recent advances in molecular genetics have led to a better understanding of the biological underpinnings of skin cancer formation. As with most cancers, the RB, p53, and RAS pathways appear to play prominent roles in the pathogenesis of several skin cancer types. Although various components of these pathways may be differentially altered in squamous cell carcinoma (SCC), basal cell carcinoma (BCC), and cutaneous melanoma, the final biochemical expression of these defects may be the same. With the unraveling of these genetic mechanisms, a more targeted approach to diagnosis and treatment may be possible in the near future.
Collapse
|
286
|
Abstract
Most current research on human brain tumors is focused on the molecular and cellular analysis of the bulk tumor mass. However, evidence in leukemia and more recently in solid tumors such as breast cancer suggests that the tumor cell population is heterogeneous with respect to proliferation and differentiation. Recently, several groups have described the existence of a cancer stem cell population in human brain tumors of different phenotypes from both children and adults. The finding of brain tumor stem cells (BTSCs) has been made by applying the principles for cell culture and analysis of normal neural stem cells (NSCs) to brain tumor cell populations and by identification of cell surface markers that allow for isolation of distinct tumor cell populations that can then be studied in vitro and in vivo. A population of brain tumor cells can be enriched for BTSCs by cell sorting of dissociated suspensions of tumor cells for the NSC marker CD133. These CD133+ cells, which also expressed the NSC marker nestin, but not differentiated neural lineage markers, represent a minority fraction of the entire brain tumor cell population, and exclusively generate clonal tumor spheres in suspension culture and exhibit increased self-renewal capacity. BTSCs can be induced to differentiate in vitro into tumor cells that phenotypically resembled the tumor from the patient. Here, we discuss the evidence for and implications of the discovery of a cancer stem cell in human brain tumors. The identification of a BTSC provides a powerful tool to investigate the tumorigenic process in the central nervous system and to develop therapies targeted to the BTSC. Specific genetic and molecular analyses of the BTSC will further our understanding of the mechanisms of brain tumor growth, reinforcing parallels between normal neurogenesis and brain tumorigenesis.
Collapse
Affiliation(s)
- Sheila K Singh
- The Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, Canada
| | | | | | | |
Collapse
|
287
|
Roussa E, Farkas LM, Krieglstein K. TGF-beta promotes survival on mesencephalic dopaminergic neurons in cooperation with Shh and FGF-8. Neurobiol Dis 2004; 16:300-10. [PMID: 15193287 DOI: 10.1016/j.nbd.2004.03.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2003] [Revised: 03/03/2004] [Accepted: 03/05/2004] [Indexed: 10/26/2022] Open
Abstract
Impaired neuronal survival is a key event in the development of degenerative diseases, such as Parkinson's disease (PD). Here we show that transforming growth factor beta (TGF-beta) acts directly on rat E14 midbrain dopaminergic neurons in vitro, its survival-promoting effect being not mediated by BDNF, NT-3, or GDNF. Treatment with TGF-beta, sonic hedgehog (Shh), or fibroblast growth factor-8 (FGF8) significantly increased number of tyrosine hydroxylase (TH)-immunoreactive neurons after 7 days, whereas application of these factors added together further increased number of TH-positive neurons, compared to single-factor treatments. Neutralization of endogenous TGF-beta, Shh, or FGF8 significantly reduced number of dopaminergic neurons. TGF-beta treatment decreased number of apoptotic cells, having no effect on cell proliferation. Neutralization of TGF-beta in vivo during chick E6-10 resulted in reduced number of midbrain dopaminergic neurons. The results suggest that TGF-beta is required for survival of mesencephalic dopaminergic neurons acting in cooperation with Shh and FGF8.
Collapse
Affiliation(s)
- Eleni Roussa
- Department for Neuroanatomy, Center of Anatomy, Georg-August-University Göttingen, 37075 Goettingen, Germany.
| | | | | |
Collapse
|
288
|
Olsen CL, Hsu PP, Glienke J, Rubanyi GM, Brooks AR. Hedgehog-interacting protein is highly expressed in endothelial cells but down-regulated during angiogenesis and in several human tumors. BMC Cancer 2004; 4:43. [PMID: 15294024 PMCID: PMC512291 DOI: 10.1186/1471-2407-4-43] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2004] [Accepted: 08/04/2004] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The Hedgehog (Hh) signaling pathway regulates a variety of developmental processes, including vasculogenesis, and can also induce the expression of pro-angiogenic factors in fibroblasts postnatally. Misregulation of the Hh pathway has been implicated in a variety of different types of cancer, including pancreatic and small-cell lung cancer. Recently a putative antagonist of the pathway, Hedgehog-interacting protein (HIP), was identified as a Hh binding protein that is also a target of Hh signaling. We sought to clarify possible roles for HIP in angiogenesis and cancer. METHODS Inhibition of Hh signaling by HIP was assayed by measuring the induction of Ptc-1 mRNA in TM3 cells treated with conditioned medium containing Sonic hedgehog (Shh). Angiogenesis was assayed in vitro by EC tube formation on Matrigel. Expression of HIP mRNA was assayed in cells and tissues by Q-RT-PCR and Western blot. HIP expression in human tumors or mouse xenograft tumors compared to normal tissues was assayed by Q-RT-PCR or hybridization of RNA probes to a cancer profiling array. RESULTS We show that Hedgehog-interacting protein (HIP) is abundantly expressed in vascular endothelial cells (EC) but at low or undetectable levels in other cell types. Expression of HIP in mouse epithelial cells attenuated their response to Shh, demonstrating that HIP can antagonize Hh signaling when expressed in the responding cell, and supporting the hypothesis that HIP blocks Hh signaling in EC. HIP expression was significantly reduced in tissues undergoing angiogenesis, including PC3 human prostate cancer and A549 human lung cancer xenograft tumors, as well as in EC undergoing tube formation on Matrigel. HIP expression was also decreased in several human tumors of the liver, lung, stomach, colon and rectum when compared to the corresponding normal tissue. CONCLUSION These results suggest that reduced expression of HIP, a naturally occurring Hh pathway antagonist, in tumor neo-vasculature may contribute to increased Hh signaling within the tumor and possibly promote angiogenesis.
Collapse
Affiliation(s)
- Catherine L Olsen
- Department of Gene Therapy, Berlex Laboratories, Inc., Richmond, CA 94806, USA
| | - Pin-Pin Hsu
- Department of Gene Therapy, Berlex Laboratories, Inc., Richmond, CA 94806, USA
- Present location: Exelixis, Inc., South San Francisco, CA 94083, USA
| | | | - Gabor M Rubanyi
- Department of Gene Therapy, Berlex Laboratories, Inc., Richmond, CA 94806, USA
| | - Alan R Brooks
- Department of Gene Therapy, Berlex Laboratories, Inc., Richmond, CA 94806, USA
| |
Collapse
|
289
|
Fan L, Pepicelli CV, Dibble CC, Catbagan W, Zarycki JL, Laciak R, Gipp J, Shaw A, Lamm MLG, Munoz A, Lipinski R, Thrasher JB, Bushman W. Hedgehog signaling promotes prostate xenograft tumor growth. Endocrinology 2004; 145:3961-70. [PMID: 15132968 DOI: 10.1210/en.2004-0079] [Citation(s) in RCA: 213] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
During fetal prostate development, Sonic hedgehog (Shh) expression by the urogenital sinus epithelium activates Gli-1 expression in the adjacent mesenchyme and promotes outgrowth of the nascent ducts. Shh signaling is down-regulated at the conclusion of prostate ductal development. However, a survey of adult human prostate tissues reveals substantial levels of Shh signaling in normal, hyperplasic, and malignant prostate tissue. In cancer specimens, the Shh expression is localized to the tumor epithelium, whereas Gli-1 expression is localized to the tumor stroma. Tight correlation between the levels of Shh and Gli-1 expression suggests active signaling between the tissue layers. To determine whether Shh-Gli-1 signaling could be functionally important for tumor growth and progression, we performed experiments with the LNCaP xenograft tumor model and demonstrated that: 1). Shh expressed by LNCaP tumor cells activates Gli-1 expression in the tumor stroma, 2). genetically engineered Shh overexpression in LNCaP cells leads to increased tumor stromal Gli-1 expression, and 3). Shh overexpression dramatically accelerates tumor growth. These data suggest that hedgehog signaling from prostate cancer cells to the stroma can elicit the expression of paracrine signals, which promote tumor growth.
Collapse
Affiliation(s)
- Lian Fan
- Oncology Group, Cambridge, Massachusetts 02163, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
290
|
Ikram MS, Neill GW, Regl G, Eichberger T, Frischauf AM, Aberger F, Quinn A, Philpott M. GLI2 is expressed in normal human epidermis and BCC and induces GLI1 expression by binding to its promoter. J Invest Dermatol 2004; 122:1503-9. [PMID: 15175043 DOI: 10.1111/j.0022-202x.2004.22612.x] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Sonic hedgehog (Shh) binds to its receptor patched (PTCH), leading to the activation and repression of target genes via the GLI family of zinc-finger transcription factors. Deregulation of the Shh pathway is associated with basal cell carcinoma (BCC) due to upregulation of GLI1 and GLI2. We recently demonstrated a positive feedback loop between GLI1 and GLI2, which revealed that GLI1 may be a direct target of GLI2. Using band shift and luciferase reporter assays, we now show that GLI2 binds the GLI-binding consensus sequence in the GLI1 promoter. These data suggest that GLI2 directly activates GLI1 and that retrovirally expressed GLI2 induces expression of endogenous GLI1 in human primary keratinocytes. Finally, using in situ hybridization, we show that GLI2 is expressed in the interfollicular epidermis and the outer root sheath of hair follicles in normal skin as well as in BCC tumor islands. These results suggest an important role for GLI2 in regulating epidermal proliferation and skin tumorigenesis.
Collapse
Affiliation(s)
- Mohammed S Ikram
- Center for Cutaneous Research, Barts and The London Queen Mary's School of Medicine & Dentistry, University of London, UK.
| | | | | | | | | | | | | | | |
Collapse
|
291
|
Lewis PM, Gritli-Linde A, Smeyne R, Kottmann A, McMahon AP. Sonic hedgehog signaling is required for expansion of granule neuron precursors and patterning of the mouse cerebellum. Dev Biol 2004; 270:393-410. [PMID: 15183722 DOI: 10.1016/j.ydbio.2004.03.007] [Citation(s) in RCA: 265] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2003] [Revised: 01/29/2004] [Accepted: 03/05/2004] [Indexed: 11/25/2022]
Abstract
The signals that promote regional growth and development of the brain are not well understood. Sonic hedgehog (Shh) is produced by Purkinje cells of the cerebellum and is a potent inducer of granule cell proliferation. Here, we demonstrate that Shh protein is present in the murine cerebellum during late stages of embryogenesis and is associated with Purkinje cell bodies and their processes. To better determine the role of Shh during cerebellar development, we genetically removed Shh activity specifically from Purkinje cells and the cerebellar anlage of the mouse embryo. We show that Shh is required for expansion of the granule neuron precursor population, but not for the subsequent differentiation of these cells. In addition, the loss of Shh activity influences Purkinje cell development and the formation of folia in the cerebellum. A role for Shh in compartmentalization of the cerebellum is also suggested by the more severe rostral defects observed in the absence of Hedgehog signaling. Together, these findings provide additional evidence for Shh's key regulatory role in controlling growth of the cerebellar primordium.
Collapse
Affiliation(s)
- Paula M Lewis
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | | | | | | | | |
Collapse
|
292
|
Abstract
Brain tumors are among the most aggressive and intractable types of cancer. Recent studies indicate that brain tumor cells resemble neural stem cells in terms of phenotype, signaling, and behavior in vitro. In light of these similarities, it has been suggested that brain tumors arise from stem cells, that they co-opt stem cell strategies for self-renewal, and even that they contain "cancer stem cells" that are critical for tumor maintenance. We will examine these possibilities and discuss their implications for the understanding and treatment of brain tumors.
Collapse
Affiliation(s)
- Trudy G Oliver
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
293
|
Berman DM, Desai N, Wang X, Karhadkar SS, Reynon M, Abate-Shen C, Beachy PA, Shen MM. Roles for Hedgehog signaling in androgen production and prostate ductal morphogenesis. Dev Biol 2004; 267:387-98. [PMID: 15013801 DOI: 10.1016/j.ydbio.2003.11.018] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2003] [Revised: 11/14/2003] [Accepted: 11/19/2003] [Indexed: 10/26/2022]
Abstract
Previous studies have demonstrated that the Hedgehog (Hh) signaling pathway plays a critical role in the development and patterning of many endodermally derived tissues. We have investigated the role of Sonic hedgehog (Shh) in formation of the prostate gland by examining the urogenital phenotype of Shh mutant fetuses. Consistent with earlier work reporting an essential role for Shh in prostate induction, we have found that Shh mutant fetuses display abnormal urogenital development and fail to form prostate buds. Unexpectedly, however, we have discovered that this prostate defect could be rescued by three different methods: renal grafting, explant culture in the presence of androgens, and administration of dihydrotestosterone (DHT) to pregnant mice, indicating that the prostate defect in Shh mutants is due to insufficient levels of androgens. Furthermore, we find that the inhibition of Hh pathway signaling by treatment with cyclopamine does not block prostate formation in explant culture, but instead produces morphological defects consistent with a role for Hh signaling in ductal patterning. Taken together, our studies indicate that the initial organogenesis of the prostate proceeds independently of Shh, but that Shh or other Hh ligands may play a role in subsequent events that pattern the prostate.
Collapse
Affiliation(s)
- David M Berman
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | | | | | | | |
Collapse
|
294
|
Ahmad I, Das AV, James J, Bhattacharya S, Zhao X. Neural stem cells in the mammalian eye: types and regulation. Semin Cell Dev Biol 2004; 15:53-62. [PMID: 15036208 DOI: 10.1016/j.semcdb.2003.09.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neural stem cells/progenitors that give rise to neurons and glia have been identified in different regions of the brain, including the embryonic retina. Recently, such cells have been reported to be present, in a mitotically quiescent state, in the ciliary epithelium of the adult mammalian eye. The retinal and ciliary epithelium stem cells/progenitors appear to share similar signaling pathways that are emerging as important regulators of stem cells in general. Yet, they are different in certain respects, such as in the potential to self-renew. These two neural stem cell/progenitor populations not only will serve as models for investigating stem cell biology but also will help explain the relationships between embryonic and adult neural stem cells/progenitors.
Collapse
Affiliation(s)
- Iqbal Ahmad
- Department of Ophthalmology, 11715 Lied Transplantation Center, University of Nebraska Medical Center, Omaha, NE 68198-7691, USA.
| | | | | | | | | |
Collapse
|
295
|
Affiliation(s)
- Marina Pasca di Magliano
- Diabetes Center, Department of Medicine, University of California-San Francisco, San Francisco, CA 94143, USA
| | | |
Collapse
|
296
|
Kim JYH, Nelson AL, Algon SA, Graves O, Sturla LM, Goumnerova LC, Rowitch DH, Segal RA, Pomeroy SL. Medulloblastoma tumorigenesis diverges from cerebellar granule cell differentiation in patched heterozygous mice. Dev Biol 2003; 263:50-66. [PMID: 14568546 DOI: 10.1016/s0012-1606(03)00434-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Medulloblastoma is a cerebellar tumor that can arise through aberrant activation of Sonic hedgehog (Shh) signaling, which normally regulates cerebellar granule cell proliferation. Mutations of the Shh receptor PATCHED (PTCH) are associated with medulloblastomas, which have not been found to have loss of PTCH heterozygosity. We address whether patched (Ptc) heterozygosity fundamentally alters granule cell differentiation and contributes to tumorigenesis by increasing proliferation and/or decreasing apoptosis in Ptc+/- mice. Our data show that postnatal Ptc+/- mouse granule cell precursor growth is not globally altered. However, many older Ptc+/- mice display abnormal cerebellar regions containing persistently proliferating granule cell precursors. Since fewer Ptc+/- mice form medulloblastomas, these granule cell rests represent a developmentally disrupted, but uncommitted stage of tumorigenesis. Although Ptc+/- mouse medulloblastomas express neurodevelopmental genes, they diverge from granule cell differentiation in their discordant coexpression of postmitotic markers despite their ongoing growth. Like human medulloblastomas, mouse tumors with reduced levels of the neurotrophin-3 receptor, trkC/Ntrk3, display decreased apoptosis in vivo, illustrating the role of TrkC in regulating tumor cell survival. These results indicate that Ptc heterozygosity contributes to tumorigenesis by predisposing a subset of granule cell precursors to the formation of proliferative rests and subsequent dysregulation of developmental gene expression.
Collapse
Affiliation(s)
- John Y H Kim
- Department of Neurology, Division of Neuroscience, Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
297
|
Abstract
The Oct-3/4 transcription factor sustains embryonic stem (ES) cell self-renewal and is a dose-dependent cell fate determinant. In the adult male, its expression is restricted to type A spermatogonia. We show that Oct-3/4 is expressed in all human testicular germ cell tumors (GCTs) tested, even in the early premalignant component. We demonstrate that Oct-3/4 dictates ES cells' oncogenic potential in a dose-dependent manner; high levels increase the malignant potential of ES cell-derived tumors while Oct-3/4 inactivation induces regression of the malignant component. Oct-3/4 expression in a heterologous cell system transforms nontumorigenic cells and endows tumorigenicity in nude mice. Our findings suggest that Oct-3/4 is not only a distinctive immunohistochemical marker for GCTs, but also plays a critical role in the genesis of these tumors.
Collapse
Affiliation(s)
- Sharon Gidekel
- Department of Experimental Medicine and Cancer Research, The Hebrew University Hadassah Medical School, Jerusalem, Israel 91120
| | | | | | | |
Collapse
|
298
|
Ruiz i Altaba A, Nguyên V, Palma V. The emergent design of the neural tube: prepattern, SHH morphogen and GLI code. Curr Opin Genet Dev 2003; 13:513-21. [PMID: 14550418 DOI: 10.1016/j.gde.2003.08.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The Sonic hedgehog (Shh) pathway plays an important role in the development of many tissues and organs. The secreted ligand Shh has been shown to act as a mitogen, morphogen and survival factor in different contexts whereas the three Gli transcription factors act as Shh mediators in a context-dependent combinatorial fashion. The common wisdom has been that Gli protein function is subject to Shh signaling. One can ask how Gli proteins act and what the nature of Shh signaling during CNS dorsal-ventral patterning is. Is it possible that Hedgehog signals are only one of several ways to regulate Gli activity? Moreover, in light of the partial rescue of the neural tube phenotype of Shh or Smoothened mutant embryos in Shh(-/-);Gli3(-/-), Smoothened(-/-);Gli3(-/-), and Shh(-/-);Rab23(-/-) double null embryos, one can consider the roles that the Shh-Gli pathway may have taken to orchestrate congruent prepattern and growth, and the importance of creating the correct number of precursors in patterning mechanisms.
Collapse
Affiliation(s)
- Ariel Ruiz i Altaba
- The Skirball Institute, New York University School of Medicine, New York, New York 10016, USA.
| | | | | |
Collapse
|
299
|
Echevarría D, Vieira C, Gimeno L, Martínez S. Neuroepithelial secondary organizers and cell fate specification in the developing brain. ACTA ACUST UNITED AC 2003; 43:179-91. [PMID: 14572913 DOI: 10.1016/j.brainresrev.2003.08.002] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In vertebrates, elaborate cellular interactions regulate the establishment of the complex structural pattern of the developing central nervous system. Distinct neural and glial identities are acquired by neuroepithelial cells, through progressive restriction of histogenetic potential under the influence of local environmental signals. The localization of the sources of such morphogenetic signals in discrete domains of the developing neural primordium has led to the concept of secondary organizers which refine the identity and polarity of neighboring neuroepithelial regions. Thus, these organizers, secondary to those that operate throughout the embryo during gastrulation, act to pattern the anterior neural plate and tube giving rise to the forebrain, midbrain and hindbrain vesicles. Important progress has recently been made in understanding their genesis and function.
Collapse
Affiliation(s)
- Diego Echevarría
- Fac. de Medicina, Instituto de Neurociencias UMH-CSIC, University Miguel Hernandez, Carretera de Valencia, N-332, Km 87, E-03550, San Juan Alicante, Spain.
| | | | | | | |
Collapse
|
300
|
Machold R, Hayashi S, Rutlin M, Muzumdar MD, Nery S, Corbin JG, Gritli-Linde A, Dellovade T, Porter JA, Rubin LL, Dudek H, McMahon AP, Fishell G. Sonic hedgehog is required for progenitor cell maintenance in telencephalic stem cell niches. Neuron 2003; 39:937-50. [PMID: 12971894 DOI: 10.1016/s0896-6273(03)00561-0] [Citation(s) in RCA: 533] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To directly test the requirement for hedgehog signaling in the telencephalon from early neurogenesis, we examined conditional null alleles of both the Sonic hedgehog and Smoothened genes. While the removal of Shh signaling in these animals resulted in only minor patterning abnormalities, the number of neural progenitors in both the postnatal subventricular zone and hippocampus was dramatically reduced. In the subventricular zone, this was partially attributable to a marked increase in programmed cell death. Consistent with Hedgehog signaling being required for the maintenance of stem cell niches in the adult brain, progenitors from the subventricular zone of floxed Smo animals formed significantly fewer neurospheres. The loss of hedgehog signaling also resulted in abnormalities in the dentate gyrus and olfactory bulb. Furthermore, stimulation of the hedgehog pathway in the mature brain resulted in elevated proliferation in telencephalic progenitors. These results suggest that hedgehog signaling is required to maintain progenitor cells in the postnatal telencephalon.
Collapse
Affiliation(s)
- Robert Machold
- Developmental Genetics Program and Department of Cell Biology, The Skirball Institute of Biomolecular Medicine, New York University Medical Center, 540 First Avenue, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|